Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Oct 1.
Published in final edited form as: Nat Rev Microbiol. 2018 Oct;16(10):594–600. doi: 10.1038/s41579-018-0039-7

Zika virus vaccines

Peter Abbink 1, Kathryn E Stephenson 1,2, Dan H Barouch 1,2,*
PMCID: PMC6162149  NIHMSID: NIHMS983945  PMID: 29921914

Abstract

The recent epidemic of Zika virus (ZIKV) in the Americas has revealed the devastating consequences of ZIKV infection, particularly in pregnant women. Congenital Zika syndrome, characterized by malformations and microcephaly in neonates as well as developmental challenges in children, highlights the need for the development of a safe and effective vaccine. Multiple vaccine candidates have been developed and have shown promising results in both animal models and phase I clinical trials. However, important challenges remain for clinical development of these vaccines. In this Progress, we discuss recent preclinical studies and lessons learned from first in-human clinical trials with ZIKV vaccines.

Table of contents blurb

The recent epidemic of Zika virus (ZIKV) in the Americas has revealed the devastating consequences of ZIKV infection, particularly in pregnant women. In this Progress, Barouch and colleagues discuss recent preclinical studies and the lessons learned from first in-human clinical trials with ZIKV vaccines.

Introduction

Zika virus (ZIKV), a flavivirus of the family Flaviviridae, was first isolated in 1947 in the Zika forest in Uganda1. ZIKV is an enveloped, positive-sense single-stranded RNA virus. Its 11kb genome encodes a single polyprotein which is cleaved into individual proteins. Structural proteins capsid (C), precursor-membrane (prM) — which is cleaved to the mature membrane protein (M) — and envelope (ENV) are assembled in virus particles (Figure 1). The non-structural proteins NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5 are involved in replication and control host cell processes to favor virus production. Until recently, infection with ZIKV was generally regarded as a self-limited, mild illness with rash, headache, myalgia and conjunctivitis, and few ZIKV infections were reported globally2. In 2007, ZIKV was recognized as the cause of an outbreak in the Yap Islands, Federated States of Micronesia3 followed in 2013 by an outbreak in French Polynesia4 before spreading to the Americas in 20155 via Easter Island, Chile6. As a result of the sudden rise in congenital abnormalities and occurrences of Guillain-Barré syndrome, the scientific community established a causal association between ZIKV infection and these neurological adverse outcomes79. This led the WHO to declare ZIKV and its suspected link to birth defects10,11 a Public Health Emergency of International Concern in February 201612.

Figure 1.

Figure 1.

The Zika virus particle and genome. Structural proteins Capsid (CAP), premembrane (prM), Envelope (ENV) and Non Structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5) are flanked by 5’ and 3’ untranslated regions (UTRs). Zika virus packages a single positive-strand RNA copy in an enveloped virus particle assembled by the structural genes. The non-structural genes are involved in viral replication and immune evasion. The primary target of neutralizing antibodies is the envelope, which together with the membrane protein is properly folded to display binding epitopes.

Research on this virus then markedly increased 1316. Studies resolved structures of the virion and the proteins that contribute to pathogenicity 1720 and defined candidate entry receptors and cell tropism21,22. Neuroprogenitor cells have been described as a preferred target for ZIKV, leading to apoptosis of these cells and congenital Zika syndrome (CZS), including microcephaly and other brain malformations23,24. The AXL receptor (also known as Tyrosine-protein kinase receptor UFO)2527, which is highly expressed on human radial glial cells, astrocytes and microglia in the developing human cortex, has been hypothesized to account for the observed neurotropism and the related congenital malformations. However, the role of AXL as an entry receptor for ZIKV remains unknown28.

The close relationship between ZIKV and other well-studied flaviviruses, such as West Nile virus (WNV), Japanese encephalitis virus (JEV), dengue virus (DENV) and tick-borne encephalitis virus (TBEV), has facilitated ZIKV research and the development of vaccines2931. Experience gained over 2 decades of research on these flaviviruses guided vaccine design and suggested protection against ZIKV may be achieved by antibodies that bind ENV25. Currently, several vaccine candidates are under development (Table 1). These include DNA vaccines, purified inactivated viruses (PIVs), live attenuated viruses (LAVs), mRNA vaccines and viral vectored vaccines (modified Vaccinia virus Ankara (MVA), measles virus (MV) and adenovirus vectors (Ad)). These efforts from multiple laboratories have led to the unprecedented pace of ZIKV vaccine development.

Table 1. Zika virus vaccines in pre-clinical and clinical development.

NAbs, neutralizing antibodies; ZPIV, ZIKV purified inactivated virus vaccine; Ad, adenovirus-based vaccine; MVA, modified Vaccinia virus Ankara; MV, measles virus-based vaccine; NA, not applicable; NS1, non-structural protein 1; ZIKV-LAV, live-attenuated vaccine; prM-ENV, precursor membrane envelope; ?, not reported or published

Vaccine Antigen Induction of NAbs Short-term protection in mice Immunocompetentce mouse model Short-term protection in monkeys Long-term protection in monkeys Advanced to clinical trial Refs
ZPIV NA Yes Yes Competent 100% 79% Phase I Larocca et al. Nature, 20167
Abbink et al. Science, 201668
Abbink et al. Sci Trans Med, 201769
DNA prM-ENV Yes Yes Competent 100% 29% Phase I/II Larocca et al. Nature, 2016
Abbink et al. Science, 2016
Dowd et al. Science, 201670
Abbink et al. Sci Trans Med, 2017
Ad prM-ENV Yes Yes Competent

Competent/Deficient
100% 100% Phase I Abbink et al. Science, 2016
Abbink et al. Sci Trans Med, 2017
Xu et al. J Virol, 201871
mRNA prM-ENV Yes Yes Competent/Deficient
Competent
100% ? Phase I/II Richner et al. Cell, 201754
Pardi et al. Nature, 201755
MVA NS1 Yes Yes Competent ? ? Phase I Brault et al. Sci Rep, 201773
MeV prM-ENV Yes ? ? ? ? Phase I www.zikavax.eu
ZIKV-LAV NA Yes Yes Competent
Deficient
Competent/Deficient
? ? NA Shan et al. Nat Med, 201774
Kwek et al. Nat Comm, 201875
Li et al, Nat Comm, 201876

In this Progress we discuss recent advances in animal models and the results from first-in-human phase I clinical trials of ZIKV vaccine candidates. In addition, we address potential challenges for late stage development of ZIKV vaccine candidates.

CZS and developmental problems

With the rapid spread of ZIKV through the Americas, many detrimental effects on fetuses and neonates were observed following infection in pregnant women32,33. In Brazil, potential confounders, such as the insecticide pyriproxyfen and the tetanus, diphtheria and pertussis (Tdap) vaccine did not correlate with the increased incidence of birth defects, whereas ZIKV confirmed by RT-PCR or antibody detection did correlate, suggesting that ZIKV was the causative agent of CZS8,34. Furthermore, animal studies have shown that ZIKV infection impacts fetal development35,36. Moreover, severe developmental problems have been observed in follow-up studies of children born with microcephaly to women confirmed to have been infected with ZIKV during pregnancy37. Developmental problems are also likely to occur in children infected during pregnancy without microcephaly, although detailed studies have not yet been completed3840.

The confirmation of ZIKV as the causative agent for CZS, combined with the severe developmental problems of neonates born with CZS, emphasizes the urgent need for a preventative vaccine. Lessons learned from congenital rubella syndrome further support that an effective vaccine might drastically reduce the incidence of infection and prevent birth defects41. However, until a vaccine is available, education and other preventative measures need to be implemented to prevent CZS42, including the development of antiviral medications43,44.

Vaccines in clinical trials

Several vaccine candidates have undergone successful pre-clinical development. Neutralizing antibodies were induced for all vaccines tested in mice (Table 1). All vaccines were able to provide short-term protection in mice against challenge with ZIKV. To date, DNA vaccines, mRNA vaccines, purified inactivated virus vaccines, and adenovirus-based vaccines have also conferred protection in monkeys. There has also been rapid advancement of these candidates into phase 1 clinical trials45. To date, there are 13 open clinical trials testing a range of ZIKV vaccine concepts, including DNA vaccines, mRNA vaccines, PIV vaccines and viral vector-based vaccines (Table 2).

Table 2.

Phase I clinical trial seroconversion rates, neutralizing antibody titers with respective assays and adoptive transfer results in mice. MN50, microneutralization assay; RVP, reporter virus particle assay; NAb, neutralizing antibody; ZPIV, ZIKV purified inactivated virus vaccine; NA, not applicable; N/S, needle and syringe; IM, intramuscular; BIDMC, Beth Israel Deaconess Medical Center; WRAIR, Walter Reed Army Institute of Research; SLU, Saint Louis University; VRC, Vaccine Research Center

Vaccine Dose Schedule NAb seroconversion MN50 titer RVP titer Adoptive transfer in mice
ZPIV (BIDMC) 5 μg Day 0 and 29, IM 10/10 (100%) 1061·7 (425·8–2489·2) NA Yes; 41/50 mice had undetectable viremia
ZPIV (WRAIR) 5 μg Day 0 and 29, IM 17/20 (85%) 100·8 (39·7–255·7) NA No
ZPIV (SLU) 5 μg Day 0 and 29, IM 25/25 (100%) 345·6 (166·4–718·0) NA No
DNA (VRC5288) 4 mg Weeks 0 and 8 by a single N/S dose 12/20 (60%) NA 67 (40 to 114) No
DNA (VRC5288) 4 mg Weeks 0 and 12 by a single N/S dose 15/20 (75%) NA 55 (39 to 78) No
DNA (VRC5288) 4 mg Weeks 0, 4, and 8 by a single N/S dose 16/20 (80%) NA 81 (51 to 127) No
DNA (VRC5288) 4 mg Weeks 0, 4, and 20 by a single N/S dose 17/19 (89%) NA 120(73 to 197) No
DNA (VRC5283) 4 mg Weeks 0, 4, and 8 by a single N/S dose 10/13 (77%) NA 48 (28 to 83) No
DNA (VRC5283) 4 mg Weeks 0, 4, and 8 by a split N/S dose 14/15 (93%) NA 150 (99 to 226) No
DNA (VRC5283) 4 mg Weeks 0, 4, and 8 by a split needle-free dose 14/14 (100%) NA 304 (215 to 430) No
DNA (Inovio Pharmaceuticals Inc) 1 mg Weeks 0, 4 and 12 by injection and followed by electroporation 12/20 (60%) 1:18 to 1:317 NA Yes; 92% survival against lethal challenge in Ifnar knockout mice
DNA (Inovio Pharmaceuticals Inc) 2 mg Weeks 0, 4 and 12 by injection and followed by electroporation 12/19 (63%) 1:18 to 1:317 NA Yes, 92% survival against lethal challenge in Ifnar knockout mice

DNA vaccines

DNA vaccines are plasmids encoding a transgene of interest under control of a promotor. DNA vaccines can be developed and produced rapidly, and they can induce both humoral and cellular immune responses46. The first clinical assessment of safety and immunogenicity of a ZIKV DNA vaccine, expressing the ZIKV precursor membrane and envelope genes (prM-ENV), was led by GeneOne Life Science, Inc and Inovio Pharmaceuticals, Inc (clinical trial NCT02809443)47. A total of 40 participants were divided equally between 2 groups and received either a 1 mg or 2 mg dose of the GLS-5700 DNA vaccine by intradermal injection with electroporation at baseline, with boosts at week 4 and week 12 (Table 2). The vaccine was well tolerated with no severe adverse reactions related to the vaccine. ZIKV-specific antibody levels at week 14 were assessed by ELISA and showed 100% seroconversion for binding antibodies in both dose groups, with a geometric mean titer (GMT) of 1,642 (347–7,760) for the 1 mg dose group and a GMT of 2,871 (705–11,688) for the 2 mg dose group. These results indicated that the vaccine-induced antibody responses were dose dependent. Neutralizing antibody titers above the detection limit were detected in 60% and 63% of the 1 mg and 2 mg dose groups, respectively. Passive transfer of week 14 serum into interferon α/β receptor (Ifnar) knockout mice followed by a lethal challenge of ZIKV48 resulted in 92% survival of mice that was independent of neutralizing antibody titer. This phase I clinical trial showed that the DNA vaccine was safe and well tolerated, and that vaccine-induced antibodies were able to protect mice from a lethal challenge of ZIKV.

Clinical trials with DNA vaccines have also been conducted by the Vaccine Research Center (VRC) of the National Institute of Allergy and Infectious Diseases (NIAID). The first DNA vaccine was designed to express ZIKV prM-ENV with a JEV envelope stem region; the JEV stem was added to increase sub-virus particle formation (vaccine VRC5288 and study VRC319; clinical trial NCT02840487). The second DNA vaccine expressed wild-type ZIKV prM-ENV (vaccine VRC5283 and study VRC320; clinical trial NCT02996461)49. In study VRC319, participants received 4 mg doses at 0 and 8 weeks, 0 and 12 weeks, 0, 4, and 8 weeks, or 0, 4, and 20 weeks by intra-muscular injection (Table 2). In VRC320, participants received 4 mg doses at 0, 4, and 8 weeks through intra-muscular injection or split-dose needle and syringe,or needle-free injection with the Stratis device50. Only mild to moderate vaccine-associated adverse events were reported. Neutralizing antibody responses were highest at 4 weeks post final vaccination. In study VRC319, neutralizing antibody GMT titers were 120 (73 to 197) with detectable neutralizing antibodies in 89% of the participants. In study VRC320, neutralizing antibody responses were detected in 100% of participants of the split-dose, needle-free delivery group, with neutralizing antibody GMTs of 304 (215 to 430). Both trials showed the DNA vaccines were well tolerated and immunogenic. The immunogenicity of the wild-type ZIKV prM-ENV DNA vaccine was higher than with the DNA vaccine that included the JEV envelope stem. The VRC5283 vaccine recently advanced into a phase II efficacy trial in regions endemic for ZIKV transmission in South and Central America, the Caribbean and the United States (NCT03110770).

Purified inactivated virus vaccines

Three ZIKV purified inactivated virus vaccine (ZPIV) phase I clinical trials (NCT02963909, NCT02952833, and NCT02937233) were reported as a combined interim analysis of the preliminary results for the identical group for each individual study51. These studies (Table 2) were conducted at Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, United States; Saint Louis University, Saint Louis, MO, United States; and Beth Israel Deaconess Medical Center (BIDMC), Boston, MA, United States. ZPIV contains a chromatographic-column-purified, formalin-inactivated ZIKV strain (PRVABC59) that was grown in Vero cells. The interim analysis included the group from each study that received the 2 dose regimen of 5 µg aluminum hydroxide adjuvanted ZPIV vaccine, administered intra-muscularly on day 1 and day 29. Adverse events related to the vaccine were mild to moderate with no serious adverse events related to the vaccine reported. Neutralizing antibody titers were determined by microneutralization (MN50) assays at WRAIR for all three trials. A total of 95% of participants had peak neutralizing antibody titers with a GMT of 286 (170–481) after the second dose. Adoptive transfer studies with purified immunoglobulin G resulted in complete protection against ZIKV challenge in 41 out of 50 Balb/c mice, and reduced viremia in the mice that were infected. Results from these trials showed that the inactivated ZIKV vaccine was well tolerated and immunogenic, and that vaccine-induced antibodies were protective in adoptive transfer studies in mice. The impact of different doses and immunization schedules will be determined in follow-up analyses of the completed studies.

Another phase I clinical trial with a ZIKV inactivated vaccine (TAK-426) led by Takeda Pharmaceutical Company Ltd, is ongoing (NCT03343626). A dose escalation study in 240 healthy individual will assess the safety and immunogenicity of this vaccine candidate.

mRNA vaccines

A newer class of vaccines, mRNA vaccines52, has also been developed against ZIKV. mRNA vaccines encode a gene of interest under control of a promotor. As mRNA is directly translated into protein after entering the cell cytoplasm, mRNA vaccines bypass the need to traverse the nuclear envelope to be expressed. This pathway could potentially lower the doses needed for mRNA vaccines while retaining the immunogenicity observed with DNA vaccines. ZIKV prM-ENV mRNA was encapsulated in a lipid nanoparticle for delivery and stability53, and immunization of both mice and monkeys with this vaccine induced high levels of neutralizing antibodies that protected against ZIKV infection54,55. In mouse pregnancy models, these mRNA vaccines prevented fetal demise, whereas fetal resorption was observed in non-immunized infected pregnant mice. However, levels of ZIKV RNA could still be detected in maternal spleen and brain as well as in placenta and in the fetal head in immunized mice56.As these results were obtained in the immunocompromised mouse model, which supports increased viral replication, it remains to be determined if viral replication would be observed in immunocompetent animal models. The first-in-human, phase I/II clinical trial led by Moderna Therapeutics is currently ongoing to assess the safety and immunogenicity of escalating doses of prM-ENV mRNA (NCT03014089) (Table 1). mRNA vaccines could be cost effective as a large number of doses can be produced efficiently. However, it remains to be determined if the promising pre-clinical data translates to humans. Additionally, the stability of mRNA vaccines needs to be taken into consideration.

Viral vector-based vaccines

Viral vector-based vaccines are another promising approach to immunize against various pathogens. These vaccines induce high humoral and cellular immune responses that have been shown to lead to protection against infection in pre-clinical models57,58. A MV ZIKV vaccine developed by Themis Bioscience GmbH is currently in a phase I clinical trial. The MV Schwarz vaccine strain59 was engineered to express ZIKV prM-ENV (MV-ZIKA) and was tested for immunogenicity in mice and monkeys60. The ongoing clinical trial is assessing safety and immunogenicity of a high or low dose when given as a single or two dose regimens (NCT02996890). An adenovirus serotype 26 (Ad26) ZIKV based vaccine (Ad26.ZIKV.001), expressing the identical antigen as the rhesus adenovirus serotype 52 (RhAd52) preclinical vaccine candidate, sponsored by Janssen Vaccines and Prevention B.V., is currently also in a phase I clinical trial (NCT03356561). This study aims to test the safety and immunogenicity of two different doses of the vaccine in a double-blind, placebo-controlled clinical trial at two sites in Kansas and Boston, United States. Experience that has already been gained with the Ad26 vaccine vector in clinical trials for other pathogens6164 may facilitate the advancement of this vaccine candidate.

Viral vector-based vaccines have shown promising results in pre-clinical models for ZIKV, and certain vectors benefit from prior experience in clinical trials for other pathogens62,65. Development of additional vectors with minimal to no pre-existing immunity is also in progress66.

Protection in pre-clinical models

At the height of the ZIKV epidemic in Brazil, multiple laboratories started to develop vaccine candidates and animal models to assess vaccine efficacy. For example, ZIKV infection in wild type BALB/c mice and rhesus monkeys largely recapitulated the magnitude and duration of ZIKV viremia in humans, exhibiting 7 to 10 days of viremia with minimal clinical symptoms. In contrast, ZIKV infection in immunodeficient mice, such as type I or I/II interferon (A129 or AG129) knockout mice or signal transducer and activator of transcription 2 (Stat2) knockout mice, were shown to exhibit prolonged viremia and have been used to study neurological disease in adult and fetal mice.

The protective efficacy of a purified inactivated Zika virus vaccine (ZPIV) and a DNA vaccine was first demonstrated in mice67. Moreover, ZPIV, a DNA vaccine, and a rhesus adenovirus serotype 52 (RhAd52) vector-based vaccine expressing a modified M-ENV ZIKV antigen were shown to block ZIKV infection in rhesus monkeys68. Furthermore, the RhAd52-based vaccine was found to provide durable complete protection in rhesus monkeys against ZIKV a year after immunization with remarkably stable neutralizing antibody titers69.

Studies from a number of groups have reported the protective efficacy of inactivated virus vaccines, DNA vaccines, viral vector-based vaccines and mRNA vaccines5456,68,7076 (Table 1). DNA vaccines expressing variations of the prM-ENV antigen were quickly developed and tested successfully for efficacy in both mice and monkeys70. mRNA vaccines expressing wild-type or modified prM-ENV antigens, leading to the generation of sub-viral particles were able to protect mice with a single dose as low as 10 µg54 or 50 µg for monkeys55. Several live-attenuated vaccines have also been developed, based on the Yellow Fever Virus (YFV) YF17D model, the JEV vaccine SA14–14-2 backbone, or attenuated through systematic deletions in the 3′UTR region in the ZIKV genome7476. All live-attenuated ZIKV vaccines have proved immunogenic and protective in mice and monkeys. Finally, MVA and MV vectors have been engineered to express the NS1 or prM-ENV proteins of ZIKV, respectively. Protection in pre-clinical models with these candidates has also been reported73.

The consistent finding from these studies is that protection against ZIKV infection is predominantly antibody-mediated. Several assays are available to measure vaccine-induced antibodies. The observation that protection is antibody-mediated is concordant with the antibody-based protection observed for WNV, JEV and DENV7779. Data suggest that titers of neutralizing antibodies of ~100, as measured by MN50 assays, are protective against ZIKV69. The plaque-reduction neutralization test (PRNT) and a ZIKV reporter viral particle assay (RVP) are other methods that are commonly used to measure neutralizing antibodies55,70. Titers between the assays vary, with the RVP reportedly being more sensitive and yielding approximately 10-fold higher titers70. CD4+ and CD8+ T cell responses may not be required if levels of neutralizing antibodies exceed this protective threshold67. However, CD8+ T cells induced by ZIKV or DENV infection have been shown to be able to reduce ZIKV viral burden in mice80,81 and further research is needed to determine the impact on short-term and long-term protection.

Protection in pregnancy

ZIKV infection in pregnant women is distinct from infection in non-pregnant women and men8284. For example, more extended periods of viremia have been observed in pregnant women and fetuses85, and ZIKV RNA was detected throughout the mother and the fetus in animal models8688. Immune responses of pregnant monkeys and mice infected with ZIKV appear similar compared to non-pregnant infected animals87,89. The ability of ZIKV to cross the fetal-placental barrier and cause damage to the fetus emphasizes the need for a vaccine and highlights the primary goal of vaccination, that is, to prevent CZS. Therefore, it will be important to measure the efficacy of vaccines to prevent fetal malformations. There are a number of aspects to this research that will need to be considered. For example, is sterilizing immunity required for efficacy or is reducing viral replication sufficient? Additionally, do immune correlates established in non-pregnant animals translate to pregnant animals?

The development of immunodeficient mouse pregnancy models for ZIKV infection has led to important advances due to the increased viral replication and impact on the central nervous system89,90, and the first prevention of fetal malformations and demise was observed using mRNA and live-attenuated ZIKV vaccines56,76,91. However, even though a statistically significant impact on fetal demise was observed, the protection was not sterilizing. ZIKV RNA was still detected in the maternal brain and spleen as well as in the placenta and fetal heads in the majority of animals. Interestingly, pups born to mothers vaccinated with a LAV were protected against lethal intracranial challenge with ZIKV76. Further research is needed to assess the impact of low level viremia in fetal and maternal compartments. In addition, with the increasing knowledge on the long term impact on children born without microcephaly but with confirmed ZIKV infection during pregnancy in humans, it is too early to tell if sterilizing immunity is required to prevent all long term sequela92. As a result of differences between rodents and primates93, a monkey pregnancy model would be preferred94 and initial progress has been reported87,95. In both rhesus and pigtail monkeys, efficient transmission of ZIKV from mother to child during pregnancy has been observed. ZIKV viremia could be detected in various anatomic compartments in the mothers and fetuses, including the brain and placenta87,95. In addition, anomalies to the brains of the fetuses were detected, ranging from white matter hypoplasia to pathology to optic nerve and eyes. Similarly as seen in humans, detrimental effects were more evident when infection occurred in early pregnancy96,97.

It is important to consider that the primary goal of a ZIKV vaccine is to prevent CZS. To realize this goal, vaccines considered for clinical development should ideally be assessed for protective efficacy in preclinical pregnancy models.

Challenges for clinical trials

With the current reduction in ZIKV transmission98,99, a phase III clinical efficacy trial could prove challenging to execute. Further development of animal pregnancy models that can effectively assess protective efficacy against CZS may therefore be important. In addition, ongoing discussions on alternative paths to licensure are being explored. Human challenge clinical trials have been conducted for other diseases such as typhoid fever100 and influenza101; however, human challenge studies for ZIKV have raised ethical discussions102. Invocation of the Food and Drug Administration (FDA) Animal Efficacy Rule (also known as the Animal Rule) could also be considered, if strong correlations of protection in pre-clinical models are deemed likely to translate to humans as outlined below104. Recently, a vaccine against anthrax was the first vaccine that was approved based on the Animal Rule103,104. According to the FDA, the Animal Rule can be pursued only if human efficacy studies cannot be performed, for example, because the conduct of such trials is unethical or because field trials after an accidental or deliberate exposure are not feasible105. For the Animal Rule to apply, the results of well-controlled animal studies need to demonstrate that clinical benefits in humans would likely be observed with the same study products. For vaccines, a clear immune correlate would facilitate the use of the Animal Rule. For ZIKV, the correlate of protection in nonhuman primates appears to be neutralizing antibody titers69.

Conclusions and perspectives

In summary, several ZIKV vaccine candidates have been shown to be safe, well-tolerated and immunogenic in humans. In the majority of trial participants, neutralizing antibody titers were induced that were comparable to titers shown to be protective in pre-clinical models. In addition, development of animal models to test vaccine efficacy in the prevention of CZS is underway.

The remarkable speed with which ZIKV vaccines have been developed has led to a rapid increase in our understanding of this virus. Nevertheless, important challenges remain for conducting clinical efficacy trials and vaccine licensure. Because of CZS and the potential lifelong impact on children born from mothers infected with ZIKV, a vaccine is urgently needed. The Rubella vaccine highlights that prevention of congenital defects can be achieved and similar success may be possible for ZIKV.

Acknowledgements

The authors like to thank Ashley York for assistance with preparing the manuscript.

Footnotes

Competing interests statement

P.A. and D.H.B. are co-inventors on ZIKV vaccine patents that have been licensed to Janssen Vaccines & Prevention B.V..

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Reviewer information

Nature Reviews Microbiology thanks Jonathan Miner, Pei-Yong Shi and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Subject categories

Biological sciences / Microbiology / Vaccines

[URI /631/326/590]

Biological sciences / Immunology / Infectious diseases

[URI /631/250/255]

Biological sciences / Microbiology / Virology

[URI /631/326/596]

References

  • 1.Dick GW, Kitchen SF & Haddow AJ Zika virus. I. Isolations and serological specificity. Trans R Soc Trop Med Hyg 46, 509–520 (1952). [DOI] [PubMed] [Google Scholar]
  • 2.Petersen LR, Baden LR, Jamieson DJ, Powers AM & Honein MA Zika Virus. New England Journal of Medicine 374, 1552–1563, doi: 10.1056/NEJMra1602113 (2016). [DOI] [PubMed] [Google Scholar]
  • 3.Duffy MR et al. Zika Virus Outbreak on Yap Island, Federated States of Micronesia. New England Journal of Medicine 360, 2536–2543, doi: 10.1056/NEJMoa0805715 (2009). [DOI] [PubMed] [Google Scholar]
  • 4.Oehler E et al. Zika virus infection complicated by Guillain-Barré syndrome – case report, French Polynesia, December 2013. Eurosurveillance 19, doi: 10.2807/1560-7917.es2014.19.9.20720 (2014). [DOI] [PubMed] [Google Scholar]
  • 5.Kindhauser MK, Allen T, Frank V, Santhana RS & Dye C Zika: the origin and spread of a mosquito-borne virus. Bulletin of the World Health Organization 94, 675–686C, doi: 10.2471/blt.16.171082 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Tognarelli J et al. A report on the outbreak of Zika virus on Easter Island, South Pacific, 2014. Archives of Virology 161, 665–668, doi: 10.1007/s00705-015-2695-5 (2015). [DOI] [PubMed] [Google Scholar]
  • 7.Carteaux G et al. Zika Virus Associated with Meningoencephalitis. N Engl J Med 374, 1595–1596, doi: 10.1056/NEJMc1602964 (2016). [DOI] [PubMed] [Google Scholar]
  • 8.de Araújo TVB et al. Association between microcephaly, Zika virus infection, and other risk factors in Brazil: final report of a case-control study. The Lancet Infectious Diseases, doi: 10.1016/s1473-3099(17)30727-2 (2017). [DOI] [PubMed] [Google Scholar]
  • 9.de Oliveira CS & da Costa Vasconcelos PF Microcephaly and Zika virus. J Pediatr (Rio J) 92, 103–105, doi: 10.1016/j.jped.2016.02.003 (2016). [DOI] [PubMed] [Google Scholar]
  • 10.Martines RB et al. Notes from the Field: Evidence of Zika Virus Infection in Brain and Placental Tissues from Two Congenitally Infected Newborns and Two Fetal Losses - Brazil, 2015. MMWR Morb Mortal Wkly Rep 65, 159–160, doi: 10.15585/mmwr.mm6506e1 (2016). [DOI] [PubMed] [Google Scholar]
  • 11.de Oliveira WK et al. Infection-related microcephaly after the 2015 and 2016 Zika virus outbreaks in Brazil: a surveillance-based analysis. The Lancet 390, 861–870, doi: 10.1016/s0140-6736(17)31368-5 (2017). [DOI] [PubMed] [Google Scholar]
  • 12.Valladeau J et al. The monoclonal antibody DCGM4 recognizes Langerin, a protein specific of Langerhans cells, and is rapidly internalized from the cell surface. Eur J Immunol 29, 2695–2704 (1999). [DOI] [PubMed] [Google Scholar]
  • 13.Sirohi D & Kuhn RJ Zika Virus Structure, Maturation, and Receptors. The Journal of Infectious Diseases 216, S935–S944, doi: 10.1093/infdis/jix515 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Faye O et al. Molecular evolution of Zika virus during its emergence in the 20(th) century. PLoS Negl Trop Dis 8, e2636, doi: 10.1371/journal.pntd.0002636 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Cunha MS et al. First Complete Genome Sequence of Zika Virus (Flaviviridae, Flavivirus) from an Autochthonous Transmission in Brazil. Genome Announc 4, doi: 10.1128/genomeA.00032-16 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Aid M et al. Zika Virus Persistence in the Central Nervous System and Lymph Nodes of Rhesus Monkeys. Cell 169, 610–620.e614, doi: 10.1016/j.cell.2017.04.008 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kostyuchenko VA et al. Structure of the thermally stable Zika virus. Nature, doi: 10.1038/nature17994 (2016). [DOI] [PubMed] [Google Scholar]
  • 18.Song H, Qi J, Haywood J, Shi Y & Gao GF Zika virus NS1 structure reveals diversity of electrostatic surfaces among flaviviruses. Nat Struct Mol Biol 23, 456–458, doi: 10.1038/nsmb.3213 (2016). [DOI] [PubMed] [Google Scholar]
  • 19.Sirohi D et al. The 3.8 A resolution cryo-EM structure of Zika virus. Science 352, 467–470, doi: 10.1126/science.aaf5316 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Osuna CE et al. Zika viral dynamics and shedding in rhesus and cynomolgus macaques. Nature Medicine 22, 1448–1455, doi: 10.1038/nm.4206 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chen J et al. AXL promotes Zika virus infection in astrocytes by antagonizing type I interferon signalling. Nature Microbiology, doi: 10.1038/s41564-017-0092-4 (2018). [DOI] [PubMed] [Google Scholar]
  • 22.Cugola FR et al. The Brazilian Zika virus strain causes birth defects in experimental models. Nature, doi: 10.1038/nature18296 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tang H et al. Zika Virus Infects Human Cortical Neural Progenitors and Attenuates Their Growth. Cell Stem Cell 18, 587–590, doi: 10.1016/j.stem.2016.02.016 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lin M-Y et al. Zika Virus Infects Intermediate Progenitor Cells and Post-mitotic Committed Neurons in Human Fetal Brain Tissues. Scientific Reports 7, doi: 10.1038/s41598-017-13980-2 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Nowakowski Tomasz J. et al. Expression Analysis Highlights AXL as a Candidate Zika Virus Entry Receptor in Neural Stem Cells. Cell Stem Cell 18, 591–596, doi: 10.1016/j.stem.2016.03.012 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Retallack H et al. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proceedings of the National Academy of Sciences 113, 14408–14413, doi: 10.1073/pnas.1618029113 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Meertens L et al. Axl Mediates ZIKA Virus Entry in Human Glial Cells and Modulates Innate Immune Responses. Cell Reports 18, 324–333, doi: 10.1016/j.celrep.2016.12.045 (2017). [DOI] [PubMed] [Google Scholar]
  • 28.Hastings AK et al. TAM Receptors Are Not Required for Zika Virus Infection in Mice. Cell Reports 19, 558–568, doi: 10.1016/j.celrep.2017.03.058 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Putnak R et al. Development of a Purified, Inactivated, Dengue-2 Virus Vaccine Prototype in Vero Cells: Immunogenicity and Protection in Mice and Rhesus Monkeys. The Journal of Infectious Diseases 174, 1176–1184, doi: 10.1093/infdis/174.6.1176 (1996). [DOI] [PubMed] [Google Scholar]
  • 30.Monath Thomas P. et al. Chimeric Live, Attenuated Vaccine against Japanese Encephalitis (ChimeriVax‐JE): Phase 2 Clinical Trials for Safety and Immunogenicity, Effect of Vaccine Dose and Schedule, and Memory Response to Challenge with Inactivated Japanese Encephalitis Antigen. The Journal of Infectious Diseases 188, 1213–1230, doi: 10.1086/378356 (2003). [DOI] [PubMed] [Google Scholar]
  • 31.Davis BS et al. West Nile Virus Recombinant DNA Vaccine Protects Mouse and Horse from Virus Challenge and Expresses In Vitro a Noninfectious Recombinant Antigen That Can Be Used in Enzyme-Linked Immunosorbent Assays. Journal of Virology 75, 4040–4047, doi: 10.1128/jvi.75.9.4040-4047.2001 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Brasil P et al. Zika Virus Infection in Pregnant Women in Rio de Janeiro. New England Journal of Medicine 375, 2321–2334, doi: 10.1056/NEJMoa1602412 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Guillemette-Artur P, Besnard M, Eyrolle-Guignot D, Jouannic JM & Garel C Prenatal brain MRI of fetuses with Zika virus infection. Pediatr Radiol, doi: 10.1007/s00247-016-3619-6 (2016). [DOI] [PubMed] [Google Scholar]
  • 34.Mlakar J et al. Zika Virus Associated with Microcephaly. N Engl J Med 374, 951–958, doi: 10.1056/NEJMoa1600651 (2016). [DOI] [PubMed] [Google Scholar]
  • 35.Martinot AJ et al. Fetal Neuropathology in Zika Virus-Infected Pregnant Female Rhesus Monkeys. Cell, doi: 10.1016/j.cell.2018.03.019 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Cugola FR et al. The Brazilian Zika virus strain causes birth defects in experimental models. Nature 534, 267–271, doi: 10.1038/nature18296 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Satterfield-Nash A et al. Health and Development at Age 19–24 Months of 19 Children Who Were Born with Microcephaly and Laboratory Evidence of Congenital Zika Virus Infection During the 2015 Zika Virus Outbreak — Brazil, 2017. MMWR. Morbidity and Mortality Weekly Report 66, 1347–1351, doi: 10.15585/mmwr.mm6649a2 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Systematic review of neuromotor impairments in infancy following congenital Zika virus infection. Developmental Medicine & Child Neurology 59, 13–13, doi: 10.1111/dmcn.14_13511 (2017).27590923 [DOI] [Google Scholar]
  • 39.Ventura CV, Maia M, Bravo-Filho V, Gois AL & Belfort R Jr. Zika virus in Brazil and macular atrophy in a child with microcephaly. Lancet 387, 228, doi: 10.1016/S0140-6736(16)00006-4 (2016). [DOI] [PubMed] [Google Scholar]
  • 40.Schuler-Faccini L et al. Possible Association Between Zika Virus Infection and Microcephaly — Brazil, 2015. MMWR. Morbidity and Mortality Weekly Report 65, 59–62, doi: 10.15585/mmwr.mm6503e2 (2016). [DOI] [PubMed] [Google Scholar]
  • 41.Plotkin SA Rubella eradication. Vaccine 19, 3311–3319, doi: 10.1016/s0264-410x(01)00073-1 (2001). [DOI] [PubMed] [Google Scholar]
  • 42.van Wouwe JP et al. Women’s reproductive health knowledge, attitudes and practices in relation to the Zika virus outbreak in northeast Brazil. Plos One 13, e0190024, doi: 10.1371/journal.pone.0190024 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Saiz J-C & Martín-Acebes MA The Race To Find Antivirals for Zika Virus. Antimicrobial Agents and Chemotherapy 61, e00411–00417, doi: 10.1128/aac.00411-17 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Shiryaev SA et al. Repurposing of the anti-malaria drug chloroquine for Zika Virus treatment and prophylaxis. Scientific Reports 7, doi: 10.1038/s41598-017-15467-6 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Barrett ADT Zika vaccine candidates progress through nonclinical development and enter clinical trials. npj Vaccines 1, doi: 10.1038/npjvaccines.2016.23 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ferraro B et al. Clinical Applications of DNA Vaccines: Current Progress. Clinical Infectious Diseases 53, 296–302, doi: 10.1093/cid/cir334 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Tebas P et al. Safety and Immunogenicity of an Anti–Zika Virus DNA Vaccine — Preliminary Report. New England Journal of Medicine, doi: 10.1056/NEJMoa1708120 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Aliota MT et al. Characterization of Lethal Zika Virus Infection in AG129 Mice. PLoS Negl Trop Dis 10, e0004682, doi: 10.1371/journal.pntd.0004682 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gaudinski MR et al. Safety, tolerability, and immunogenicity of two Zika virus DNA vaccine candidates in healthy adults: randomised, open-label, phase 1 clinical trials. The Lancet, doi: 10.1016/s0140-6736(17)33105-7 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yousafzai MT et al. Feasibility of conducting intradermal vaccination campaign with inactivated poliovirus vaccine using Tropis intradermal needle free injection system, Karachi, Pakistan. Heliyon 3, e00395, doi: 10.1016/j.heliyon.2017.e00395 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Modjarrad K et al. Preliminary aggregate safety and immunogenicity results from three trials of a purified inactivated Zika virus vaccine candidate: phase 1, randomised, double-blind, placebo-controlled clinical trials. The Lancet, doi: 10.1016/s0140-6736(17)33106-9 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Pardi N, Hogan MJ, Porter FW & Weissman D mRNA vaccines — a new era in vaccinology. Nature Reviews Drug Discovery, doi: 10.1038/nrd.2017.243 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Reichmuth AM, Oberli MA, Jaklenec A, Langer R & Blankschtein D mRNA vaccine delivery using lipid nanoparticles. Therapeutic Delivery 7, 319–334, doi: 10.4155/tde-2016-0006 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Richner JM et al. Modified mRNA Vaccines Protect against Zika Virus Infection. Cell 168, 1114–1125.e1110, doi: 10.1016/j.cell.2017.02.017 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Pardi N et al. Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination. Nature 543, 248–251, doi: 10.1038/nature21428 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Richner JM et al. Vaccine Mediated Protection Against Zika Virus-Induced Congenital Disease. Cell 170, 273–283.e212, doi: 10.1016/j.cell.2017.06.040 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ura T, Okuda K & Shimada M Developments in Viral Vector-Based Vaccines. Vaccines 2, 624–641, doi: 10.3390/vaccines2030624 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lauer KB, Borrow R, Blanchard TJ & Papasian CJ Multivalent and Multipathogen Viral Vector Vaccines. Clinical and Vaccine Immunology 24, e00298–00216, doi: 10.1128/cvi.00298-16 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Combredet C et al. A Molecularly Cloned Schwarz Strain of Measles Virus Vaccine Induces Strong Immune Responses in Macaques and Transgenic Mice. Journal of Virology 77, 11546–11554, doi: 10.1128/jvi.77.21.11546-11554.2003 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ulmer JB et al. Heterologous protection against influenza by injection of DNA encoding a viral protein. Science 259, 1745–1749 (1993). [DOI] [PubMed] [Google Scholar]
  • 61.Barouch DH et al. Characterization of Humoral and Cellular Immune Responses Elicited by a Recombinant Adenovirus Serotype 26 HIV-1 Env Vaccine in Healthy Adults (IPCAVD 001). The Journal of Infectious Diseases 207, 248–256, doi: 10.1093/infdis/jis671 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Baden LR et al. Assessment of the Safety and Immunogenicity of 2 Novel Vaccine Platforms for HIV-1 Prevention. Annals of Internal Medicine 164, 313, doi: 10.7326/m15-0880 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Milligan ID et al. Safety and Immunogenicity of Novel Adenovirus Type 26– and Modified Vaccinia Ankara–Vectored Ebola Vaccines. Jama 315, 1610, doi: 10.1001/jama.2016.4218 (2016). [DOI] [PubMed] [Google Scholar]
  • 64.Winslow RL et al. Immune Responses to Novel Adenovirus Type 26 and Modified Vaccinia Virus Ankara–Vectored Ebola Vaccines at 1 Year. Jama 317, 1075, doi: 10.1001/jama.2016.20644 (2017). [DOI] [PubMed] [Google Scholar]
  • 65.Ledgerwood JE et al. Chimpanzee Adenovirus Vector Ebola Vaccine - Preliminary Report. N Engl J Med, doi: 10.1056/NEJMoa1410863 (2014). [DOI] [PubMed] [Google Scholar]
  • 66.Abbink P et al. Rapid Cloning of Novel Rhesus Adenoviral Vaccine Vectors. Journal of Virology 92, e01924–01917, doi: 10.1128/jvi.01924-17 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Larocca RA et al. Vaccine protection against Zika virus from Brazil. Nature 536, 474–478, doi: 10.1038/nature18952 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Abbink P et al. Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys. Science 353, 1129–1132, doi: 10.1126/science.aah6157 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Abbink P et al. Durability and correlates of vaccine protection against Zika virus in rhesus monkeys. Science Translational Medicine 9, eaao4163, doi: 10.1126/scitranslmed.aao4163 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Dowd KA et al. Rapid development of a DNA vaccine for Zika virus. Science 354, 237–240, doi: 10.1126/science.aai9137 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Xu K et al. Recombinant Chimpanzee Adenovirus Vaccine AdC7-M/E Protects against Zika Virus Infection and Testis Damage. Journal of Virology, JVI.01722–01717, doi: 10.1128/jvi.01722-17 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Larocca RA et al. Adenovirus serotype 5 vaccine vectors trigger IL-27-dependent inhibitory CD4+ T cell responses that impair CD8+ T cell function. Science Immunology 1, eaaf7643–eaaf7643, doi: 10.1126/sciimmunol.aaf7643 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Brault AC et al. A Zika Vaccine Targeting NS1 Protein Protects Immunocompetent Adult Mice in a Lethal Challenge Model. Sci Rep 7, 14769, doi: 10.1038/s41598-017-15039-8 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Shan C et al. A live-attenuated Zika virus vaccine candidate induces sterilizing immunity in mouse models. Nature Medicine 23, 763–767, doi: 10.1038/nm.4322 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kwek SS et al. A systematic approach to the development of a safe live attenuated Zika vaccine. Nature Communications 9, doi: 10.1038/s41467-018-03337-2 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Li X-F et al. Development of a chimeric Zika vaccine using a licensed live-attenuated flavivirus vaccine as backbone. Nature Communications 9, doi: 10.1038/s41467-018-02975-w (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Austin SK & Dowd KA B cell response and mechanisms of antibody protection to West Nile virus. Viruses 6, 1015–1036, doi: 10.3390/v6031015 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Xu M et al. A potent neutralizing antibody with therapeutic potential against all four serotypes of dengue virus. npj Vaccines 2, doi: 10.1038/s41541-016-0003-3 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Larena M, Prow NA, Hall RA, Petrovsky N & Lobigs M JE-ADVAX Vaccine Protection against Japanese Encephalitis Virus Mediated by Memory B Cells in the Absence of CD8+ T Cells and Pre-Exposure Neutralizing Antibody. Journal of Virology 87, 4395–4402, doi: 10.1128/jvi.03144-12 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wen J et al. Identification of Zika virus epitopes reveals immunodominant and protective roles for dengue virus cross-reactive CD8+ T cells. Nature Microbiology 2, 17036, doi: 10.1038/nmicrobiol.2017.36 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Elong Ngono A et al. Mapping and Role of the CD8 + T Cell Response During Primary Zika Virus Infection in Mice. Cell Host & Microbe 21, 35–46, doi: 10.1016/j.chom.2016.12.010 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Driggers RW et al. Zika Virus Infection with Prolonged Maternal Viremia and Fetal Brain Abnormalities. N Engl J Med, doi: 10.1056/NEJMoa1601824 (2016). [DOI] [PubMed] [Google Scholar]
  • 83.El Costa H et al. ZIKA virus reveals broad tissue and cell tropism during the first trimester of pregnancy. Scientific Reports 6, doi: 10.1038/srep35296 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Osuna CE & Whitney JB Nonhuman Primate Models of Zika Virus Infection, Immunity, and Therapeutic Development. The Journal of Infectious Diseases 216, S928–S934, doi: 10.1093/infdis/jix540 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Bhatnagar J et al. Zika Virus RNA Replication and Persistence in Brain and Placental Tissue. Emerging Infectious Diseases 23, 405–414, doi: 10.3201/eid2303.161499 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Miner Jonathan J. et al. Zika Virus Infection during Pregnancy in Mice Causes Placental Damage and Fetal Demise. Cell, doi: 10.1016/j.cell.2016.05.008 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Nguyen SM et al. Highly efficient maternal-fetal Zika virus transmission in pregnant rhesus macaques. PLoS Pathog 13, e1006378, doi: 10.1371/journal.ppat.1006378 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Li C et al. Zika Virus Disrupts Neural Progenitor Development and Leads to Microcephaly in Mice. Cell Stem Cell 19, 120–126, doi: 10.1016/j.stem.2016.04.017 (2016). [DOI] [PubMed] [Google Scholar]
  • 89.Vermillion MS et al. Intrauterine Zika virus infection of pregnant immunocompetent mice models transplacental transmission and adverse perinatal outcomes. Nature Communications 8, 14575, doi: 10.1038/ncomms14575 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Mysorekar IU, Phimister EG & Diamond MS Modeling Zika Virus Infection in Pregnancy. New England Journal of Medicine 375, 481–484, doi: 10.1056/NEJMcibr1605445 (2016). [DOI] [PubMed] [Google Scholar]
  • 91.Shan C et al. A single-dose live-attenuated vaccine prevents Zika virus pregnancy transmission and testis damage. Nature Communications 8, doi: 10.1038/s41467-017-00737-8 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.van der Linden V et al. Description of 13 Infants Born During October 2015–January 2016 With Congenital Zika Virus Infection Without Microcephaly at Birth — Brazil. MMWR. Morbidity and Mortality Weekly Report 65, 1343–1348, doi: 10.15585/mmwr.mm6547e2 (2016). [DOI] [PubMed] [Google Scholar]
  • 93.Malassine A, Frendo JL & Evain-Brion D A comparison of placental development and endocrine functions between the human and mouse model. Human Reproduction Update 9, 531–539, doi: 10.1093/humupd/dmg043 (2003). [DOI] [PubMed] [Google Scholar]
  • 94.Grigsby P Animal Models to Study Placental Development and Function throughout Normal and Dysfunctional Human Pregnancy. Seminars in Reproductive Medicine 34, 011–016, doi: 10.1055/s-0035-1570031 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Adams Waldorf KM et al. Fetal brain lesions after subcutaneous inoculation of Zika virus in a pregnant nonhuman primate. Nature Medicine 22, 1256–1259, doi: 10.1038/nm.4193 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Lissauer D, Smit E & Kilby MD Zika virus and pregnancy. BJOG, doi: 10.1111/1471-0528.14071 (2016). [DOI] [PubMed] [Google Scholar]
  • 97.Rather IA, Lone JB, Bajpai VK & Park Y-H Zika Virus Infection during Pregnancy and Congenital Abnormalities. Frontiers in Microbiology 8, doi: 10.3389/fmicb.2017.00581 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Ulmer JB, Deck RR, Dewitt CM, Donnhly JI & Liu MA Generation of MHC class I-restricted cytotoxic T lymphocytes by expression of a viral protein in muscle cells: antigen presentation by non-muscle cells. Immunology 89, 59–67 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.van Ballegooijen M, Bogaards JA, Weverling GJ, Boerlijst MC & Goudsmit J AIDS vaccines that allow HIV-1 to infect and escape immunologic control: a mathematic analysis of mass vaccination. J Acquir Immune Defic Syndr 34, 214–220 (2003). [DOI] [PubMed] [Google Scholar]
  • 100.Feasey NA & Levine MM Typhoid vaccine development with a human challenge model. The Lancet 390, 2419–2421, doi: 10.1016/s0140-6736(17)32407-8 (2017). [DOI] [PubMed] [Google Scholar]
  • 101.Memoli MJ et al. Validation of the Wild-type Influenza A Human Challenge Model H1N1pdMIST: An A(H1N1)pdm09 Dose-Finding Investigational New Drug Study. Clinical Infectious Diseases 60, 693–702, doi: 10.1093/cid/ciu924 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Ethical Considerations for Zika Virus Human Challenge Trials, <https://www.niaid.nih.gov/sites/default/files/EthicsZikaHumanChallengeStudiesReport2017.pdf> (2017).
  • 103.Beasley DWC, Brasel TL & Comer JE First vaccine approval under the FDA Animal Rule. npj Vaccines 1, doi: 10.1038/npjvaccines.2016.13 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Product Development Under the Animal Rule. Guidance for Industry, <https://www.fda.gov/downloads/drugs/guidances/ucm399217.pdf> (2015).
  • 105.A Hokey D TB Vaccines: The (Human) Challenge Ahead. Mycobacterial Diseases 04, doi: 10.4172/2161-1068.1000e128 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES